• Aucun résultat trouvé

Modeling the 3D functional architecture of the nucleus in animal and plant kingdoms

N/A
N/A
Protected

Academic year: 2021

Partager "Modeling the 3D functional architecture of the nucleus in animal and plant kingdoms"

Copied!
11
0
0

Texte intégral

(1)

HAL Id: hal-02610642

https://hal.archives-ouvertes.fr/hal-02610642

Submitted on 17 May 2020

HAL is a multi-disciplinary open access archive for the deposit and dissemination of sci- entific research documents, whether they are pub- lished or not. The documents may come from teaching and research institutions in France or abroad, or from public or private research centers.

L’archive ouverte pluridisciplinaire HAL, est destinée au dépôt et à la diffusion de documents scientifiques de niveau recherche, publiés ou non, émanant des établissements d’enseignement et de recherche français ou étrangers, des laboratoires publics ou privés.

Modeling the 3D functional architecture of the nucleus in animal and plant kingdoms

Valérie Gaudin, Philippe Andrey, Eve Devinoy, Clémence Kress, Kiên Kiêu, Nathalie Beaujean, Yves Maurin, Pascale Debey

To cite this version:

Valérie Gaudin, Philippe Andrey, Eve Devinoy, Clémence Kress, Kiên Kiêu, et al.. Modeling the

3D functional architecture of the nucleus in animal and plant kingdoms. Comptes Rendus Biologies,

Elsevier, 2009, 332 (11), pp.937-946. �10.1016/j.crvi.2009.09.001�. �hal-02610642�

(2)

C. R. Biologies 332 (2009) 937–946

Review / Revue

Modeling the 3D functional architecture of the nucleus in animal and plant kingdoms

Valérie Gaudin

a

, Philippe Andrey

b,c,g

, Eve Devinoy

d

, Clémence Kress

d

, Kiên Kieu

e

, Nathalie Beaujean

f

, Yves Maurin

b,c

, Pascale Debey

f,

aLaboratoire de biologie cellulaire, UR501, IJPB, route de Saint-Cyr, INRA, 78026 Versailles, France

bINRA, UMR1197, Neurobiologie de l’olfaction et de la prise alimentaire, Domaine de Vilvert, 78350 Jouy-en-Josas, France cUniversité Paris-Sud 11, UMR1197, 91405 Orsay, France

dINRA, UR1196 Génomique et physiologie de la lactation, Domaine de Vilvert, F-78350 Jouy-en-Josas, France eINRA, UR341, Mathématiques et informatique appliquée, Domaine de Vilvert, 78350 Jouy-en-Josas, France fINRA, UMR1198 Biologie du développement et de la reproduction, Domaine de Vilvert, 78350 Jouy-en-Josas, France

gUPMC, Univ Paris 06, France Available online 15 October 2009

Presented by Michel Thellier

Abstract

Compartmentalization is one of the fundamental principles which underly nuclear function. Numerous studies describe complex and sometimes conflicting relationships between nuclear gene positioning and transcription regulation. Therefore the question is whether topological landmarks and/or organization principles exist to describe the nuclear architecture and, if existing, whether these principles are identical in the animal and plant kingdoms. In the frame of an agroBI-INRA program on nuclear architecture, we set up a multidisciplinary approach combining biological studies, spatial statistics and 3D modeling to investigate spatial organization of a nuclear compartment in both plant and animal cells in their physiological contexts. In this article, we review the questions addressed in this program and the methodology of our work.

To cite this article: V. Gaudin et al., C. R. Biologies 332 (2009).

©

2009 Académie des sciences. Published by Elsevier Masson SAS. All rights reserved.

Résumé

Modéliser l’architecture tri-dimensionnelle du noyau dans des cellules animales et végétales.

L’organisation en comparti- ments est un des principes fondamentaux qui gouverne le fonctionnement du noyau. De nombreuses études montrent des relations complexes et parfois contradictoires entre la position d’un gène dans le noyau et son état transcriptionnel. Deux questions se posent : existe-t-il des principes généraux gouvernant l’organisation nucléaire, et, si oui, ces principes sont-ils analogues dans les règnes animal et végétal ? Dans le cadre d’un programme agroBI-INRA sur l’architecture nucléaire, nous avons développé une ap- proche multidisciplinaire combinant expériences biologiques, statistiques spatiales et modélisation en 3 dimensions pour analyser l’organisation spatiale d’un compartiment nucléaire dans des cellules animales et végétales, dans leur environnement physiolo- gique. Dans cet article, nous faisons une revue autour des questions posées dans ce programme et présentons la méthodologie de ce travail.

Pour citer cet article : V. Gaudin et al., C. R. Biologies 332 (2009).

©

2009 Académie des sciences. Published by Elsevier Masson SAS. All rights reserved.

* Corresponding author.

E-mail address:debey@mnhn.fr(P. Debey).

1631-0691/$ – see front matter ©2009 Académie des sciences. Published by Elsevier Masson SAS. All rights reserved.

doi:10.1016/j.crvi.2009.09.001

(3)

Keywords:Nuclear architecture; Heterochromatin; Chromocenter; Confocal imaging; Digital image analysis; 3D modeling; Spatial statistics Mots-clés :Architecture nucléaire ; Hétérochromatine ; Chromocentre ; Imagerie confocale ; Analyse d’images numériques ; Reconstruction 3D ;

Statistique spatiale

1. Introduction

Increasing evidence suggests that besides DNA reg- ulatory sequences and transcription factors, the spatial and temporal organization of nuclear processes (tran- scription, reparation, replication, . . . ) as well as chromo- some and gene distribution within the nuclear space are central determinants of genome functions. An impor- tant goal of the post-genomic systems biology will be therefore to better understand genome function within the architectural framework of the nucleus and integrate different levels of genome regulation [1,2]. However, deciphering the three-dimensional (3D) nuclear organi- zation in a relevant physiological context is a difficult task due to technical constraints and requirements of imaging and computational modeling.

To develop a systems biology view of the nucleus and to give standardized and statistical representations of the nuclear architecture in order to highlight pos- sible organizational rules, we investigated the spatial organization of a nuclear compartment in both plant and animal cells. We set up a multidisciplinary ap- proach combining biological studies, spatial statistics and 3D modeling in the frame of an agroBI-INRA pro- gram entitled “Nuclear Architecture: spatial modeling and application to a better understanding of differentia- tion/dedifferentiation mechanisms”. This article aims at presenting the conceptual background, the main goals and the methodology of our work.

2. Functional nuclear organization: background 2.1. Elements of the nuclear organization

Compartmentalization is one of the fundamental principles regulating organization-function crosstalk in the nucleus. As defined by Misteli, a “compartment”

is “a macroscopic region within the nucleus that is morphologically and/or functionally distinct from its surrounding” [3]. The compartments may be either sites highly enriched in a definite set of nuclear pro- teins (nucleoli, speckles, Cajal bodies, promyelocytic leukemia protein (PLM) nuclear bodies, interchromatin granules, . . . ) or chromatin domains, which we will fo- cus on (Fig. 1). One of the most studied compartments is “constitutive heterochromatin”, a major component of eukaryotic genomes usually formed by large blocks

located near centromeres and telomeres and that has a compact organization throughout cell cycle [4–6]. It is opposed to “euchromatin”, which exhibits a more

“open” conformation and corresponds to actively tran- scribed regions of the genome. Euchromatin can be turned temporarily into heterochromatin upon silencing, being thus termed “facultative heterochromatin”. Hete- rochromatin can be distinguished from euchromatin by cytological properties, compaction levels and epigenetic marks (Fig. 1). However, several studies have shown that transcription can occur in heterochromatin regions in yeast, plants and animals [7–9].

Furthermore, large-scale mapping of the human genome has shown a link between chromatin morphol- ogy and gene density, with gene-rich regions being present in open chromatin regions and gene-poor re- gions in more condensed domains, regardless of their activity status [10]. In some animal species such as mouse and, to a lesser extent human, the pericentric heterochromatin of several chromosomes assembles to form large heterochromatic domains called “chromo- centers” [11,12]. In Arabidopsis thaliana, discrete and intensely stained heterochromatic compartments are re- ferred to as chromocenters (Fig. 1). They correspond to centromeric and pericentromeric regions and were described as anchoring regions for 0.1–1.5 Mbp euchro- matin loops allowing to propose a chromocenter-loop nuclear organizational model [13,14]. Chromocenters can be visualized using FISH [13], immunolocalization [15] or GFP markers [16].

Since the first laser microbeam irradiation experi-

ments [17], it is now well demonstrated that each chro-

mosome of an animal or plant cell’s nucleus is con-

fined to a discrete region of the interphase nucleus,

referred to as a chromosome territory (CT) and corre-

sponding to a chromatin compartment at a larger scale

(Fig. 1) [18–20]. In mammalian cells, the distribution

of CT appears non-random, with most gene-rich CTs

preferentially localized in the nuclear interior, whereas

gene-poor CTs localized toward the nuclear periphery

[18,21,22]. Such arrangements are evolutionarily con-

served [23,24]. Furthermore, cell or nuclear shapes and

chromosome size have been observed to influence the

3D arrangement of CTs: for example, the ellipsoid hu-

man fibroblast nuclei present a chromosome size de-

pendent pattern even if the radial distribution of gene-

rich and gene-poor CTs was found to be recapitulated

(4)

V. Gaudin et al. / C. R. Biologies 332 (2009) 937–946 939

Fig. 1. Compartmentalization of the nucleus and elements of its architecture. A–C. Heterochromatin compartments. A. Mouse fibroblast nucleus (Hoechst staining). B. Epithelial cell nucleus from a rabbit mammary gland in lactation (tissue section, DAPI staining). C. Nucleus isolated from anA. thalianaplantlet (DAPI staining). D–F. Compartments defined as regions enriched in nuclear proteins. D. Nucleoli in an epithelial cell nucleus from a rabbit mammary gland in lactation; immunolocalization of B23 protein (green), DNA counterstained with DAPI (blue) (tissue section). E. Splicing speckles in the nucleus of a mouse mammary epithelial cell (cultured HC11 cell line); immunolocalization of SC35 protein (green), DNA counterstained with propidium iodide (red). F. Nuclear localization of LHP1, the plant homolog of Heterochromatin Protein 1 (HP1) in a root cell nucleus from a transgenic line expressing the LHP1-GFP fusion protein. G–I. Chromocenters, centromeres and telomeres.

G. FISH with a pancentromeric probe (green) and a LNA telomeric probe (red) in an HC11 cell nucleus; DNA counterstained with DAPI (blue).

H. Immunolocalization of pericentromeres (HP1 protein, red) and centromeres (CENP proteins, green) in the nucleus of a rabbit embryo at 8-cell stage. I. 5S (green) and 45S rDNA (red) repeats in anA. thalianaleaf nucleus revealed by FISH; DNA counterstained with DAPI (blue) (kindly provided by F. Tessadori). J–L. Visualization of genomic regions of different sizes by FISH. J. Localization of genes encoding casein (green) and whey acidic protein (white) in the nucleus of HC11 cells after hybridization with gene specific BAC probes and DNA staining with propidium iodide (red). K. Localization of 3 genomic regions (about 100 kbp) with 3 BAC probes (2 in green and one in red) in anA. thalianaleaf nucleus (kindly provided by F. Tessadori). L. Territory of chromosome 15 (green) in the nucleus of a cultured rabbit embryonic fibroblast; DNA counterstained with propidium iodide (red). All images were acquired on confocal microscopes.

(5)

at the sub-chromosomal level [25–27]. In A. thaliana, CTs were described as predominantly randomly located within the nucleus. Only chromosomes 2 and 4, which bear nucleolus organizer regions (NOR), were associ- ated more frequently than randomly [19,20]. The rela- tionships between CTs distribution and nuclear organi- zation require therefore further investigation. Parame- ters such as DNA content, gene density, and transcrip- tional activity of chromosomal sub-domains might all come into account and raise the question of the mecha- nism of chromosome positioning.

2.2. Relationship between gene positioning and transcriptional activity

Numerous studies described complex and sometimes conflicting relationships between gene positioning with respect to several compartments, and transcription reg- ulation.

2.2.1. Relative positioning of genes with respect to nuclear compartments

Some studies address the question of comparative positioning inside the nucleus by examining different genes in a given cell, or the same gene in different cell types.

As stated above, gene-rich chromosomes are usually more centrally located than gene-poor chromosomes in human [22], chicken [28], hydra [29], and this trend is observed also for subchromosomal domains [25,30].

The nuclear periphery, which is lined in most higher eu- karyotes by a peripheral ring of condensed chromatin, represents generally a repressive zone: some genes are preferentially associated with the nuclear periphery in cells where they are inactive, and more centrally lo- cated in cells where they are highly transcribed [31–34].

More generally, late replicating foci, assumed to corre- spond to inactive chromatin are located at the nuclear periphery. However, location of transcribed genes at the nuclear envelope has also been observed in yeast [35], possibly because of their association with proteins of the nuclear-pore complexes [36]. It is not known whether similar gene activation processes exist at the nuclear pe- riphery in higher eukaryotes. The potential role of the nuclear periphery in genome regulation has become of particular importance in view of human diseases caused by mutations in genes encoding the architectural pro- teins of the lamina [37].

The relationship between location and transcrip- tional status of genomic sequences with respect to their CTs is the subject of conflicting reports: non-coding sequences are randomly distributed or preferentially lo-

cated at the interior of the CT, whereas several genes have been found preferentially located at the periph- ery of the CT, irrespective of their transcriptional sta- tus [38,39]. This peripheral location might facilitate access to the transcriptional machinery located in the inter-chromosomal space [40,41]. Chromatin loops con- taining large gene clusters (major histocompatibility complex locus, epidermal differentiation complex) can even loop out of their CTs into these interchromosomal compartments under conditions where they are highly expressed [42,43]. On the other hand, some active genes reside in the CT interior [44], suggesting that CTs can be invaginated by infoldings of the interchromosomal compartment [41,45] (reviewed in [46]).

Proximity to centromeric heterochromatin is gener- ally associated with gene silencing, the most striking examples being the association of T-cell specific and developmentally regulated B-cell genes with the Ikaros complex at the centromeric heterochromatin [47] and the beta-globin gene in erythroid cells [48].

2.2.2. Dynamic gene repositioning upon activation/inhibition

Studying the dynamics of genes during the differ- entiation process has brought other strong arguments for a relationship between nuclear positioning and tran- scription status [3,49]. For instance, the Hoxb1 locus loops out of its CT during transcriptional activation [50]; movement towards the nuclear periphery was ob- served upon inactivation [51], or towards the nuclear in- terior upon neural induction [33]; or gene repositioning was described away of the chromocenters upon activa- tion [48] or towards the chromocenters upon silencing [52]. Finally, active genes widely separated in cis or located on different chromosomes can even colocalize possibly as a result of migration upon activation towards preassembled transcription sites [53,54]. Similarly, the ribosomal RNA genes coalesce in the nucleolus where RNA polymerase I accumulates [55]. More recently, in- terchromatin granules were suggested to actively recruit genomic loci, thus forming “enhancer hubs” for inter- chromosomal interactions [56].

2.3. Nuclear organization depends on the physiological status of the cell

Besides the above-described changes in gene loca-

tion, changes in nuclear architecture itself are amongst

the most dramatic hallmarks of cell differentiation, de-

velopment or malignant processes [26,31,49,57,58]. For

instance, condensation or morphology of CTs may vary

with differentiation in chicken, mouse, or human cells

(6)

V. Gaudin et al. / C. R. Biologies 332 (2009) 937–946 941

[59,60]. The spatial organization of centromeric hete- rochromatin in haematopoeitic cells changes with cell types and is ontogenically determined [12]. Clustering of pericentric heterochromatin has been reported during terminal differentiation, e.g. during myogenesis [61], in human neutrophils [62], human and mouse neurons [63]

and rat myoblasts [64]. In naturally fertilized mouse em- bryos, the specific radial (“cartwheel”) organization of chromosomes at 1-cell is progressively modified into a “somatic cell-type” organization during development [65,66]. Interestingly, similar 1-cell type nuclear orga- nization is adopted upon transfer of a somatic nucleus in the oocyte cytoplasm [66,67].

In plants, heterochromatin content varies with dif- ferentiation [68–70]. Peculiar heterochromatin organi- zation with smaller chromocenters and additional het- erochromatic foci interspersed in euchromatin was ob- served in endosperm nuclei [71] suggesting a relaxed chromocenter-loop organization of the nuclei. Nuclear compaction of specific loci depends on their transcrip- tional status in regards to cell differentiation [72] and changes in chromocenter and chromatin organization accompany changes in cell fate and differentiation [70,73].

2.4. Nuclear organization in natural environments Until now, most data have been gathered on isolated plant cell nuclei or on nuclei from immortalized animal cell lines outside their physiological environment, ex- cept for circulating blood cells. Little is known about nuclear organization of cells within their tissues [74].

Few studies compared nuclear organization in primary cells versus cell lines, in cell lines versus tissues, in 2D culture versus 3D cultures and suggested a control of tissue architecture over nuclear organization [75–77].

In addition, few data on nuclear organization were ac- quired in plant cell nuclei in situ [71,72]. In vivo ap- proaches allowed distinguishing three main types of plant nuclei based on their nuclear shapes (round, elon- gated and rod-like nuclei), which are associated with various cell-type or tissues [78]. Studies of chromatin dynamics in plants have shown that confinement area for tagged loci is different according to cell types [79].

Further investigation will be required in plant cell nu- clei.

2.5. Current models of nuclear organization

Despite being at a very beginning of a system-level view of the nucleus, two major models, a determinis- tic model and a self-organizing model, emerged [2].

The deterministic model proposes that structure dictates function and is consistent with stable observed struc- tures in the nucleus. However, some experimental ev- idence does not support this model, such as the loss of prime structural elements without any major conse- quences on genome function. On the other hand, the nucleus can be seen as a self-organizing system, based on the dynamic interaction of nuclear components and various interplay between structure and function with self-reinforcing mechanism. Indeed, nuclear processes can lead to nuclear changes in global architecture and de novo formation of nuclear structures. For example, sup- ported by theoretical considerations as well as several experimental results from chromosome conformation capture (3C), transcription machinery may function as a main molecular tie that maintain most genomic loops and determine gene activity [80]. 3C experiments have also recently revealed that repeat elements and nucle- olus play central roles in the dynamic organization of genome architecture [81]. However, as most probably, the two models are not exclusive and additional forces may have to be taken into considerations to explain the structure/function of the nucleus such as stochas- tic collisions [82], non-specific entropy-driven crowd- ing forces [83] or “depletion-attraction” forces, highly significant in crowded environments like those in nuclei [14,84,85].

3. Overview of the project

3.1. Aim of the project and main achievements

Two main questions arising from the existing data were at the origin of the project:

• Are there organization principles, and/or topo- graphical landmarks to describe the nuclear archi- tecture? If existing, are these principles identical in the animal and plant kingdoms?

• What transformations allow to process from one differentiation state to another? How are co-regu- lated gene networks positioned with respect to these topological landmarks?

To address these questions, we chose to analyze the

centric/pericentric heterochromatin compartment and

its three-dimensional nuclear distribution. Indeed, the

centric/pericentric compartment is well defined and

present in all cell types with some already described

variations. Its structure and cohesion are essential for

genome organization and proper cell division [70,73,

(7)

86,87] and it is involved in genome regulation, usually behaving as a transcriptional repressor compartment.

Aiming at producing generic models for the nuclear architecture in physiological environment, we produced images of 3D-preserved nuclei both from animal and plant kingdoms in three different biological systems to be able to extract robust organizational principles. We performed fluorescence or immunofluorescence label- ing to reveal the structural elements, generated a ho- mogenous 3D-image collection of cell nuclei acquired in standardized conditions (ICOPAN website/image re- source: http://amib.jouy.inra.fr/icopan/) and developed image processing and spatial statistical tools to analyze and model the spatial distribution of the studied com- partment.

3.2. Choice of the biological systems

Three biological complex systems were chosen, two animal (rabbit) systems and one plant (Arabidop- sis thaliana) system. Rabbit was chosen since: (i) its complete genome sequence (2X coverage) was already available and its 7X coverage was expected to be re- leased in a near future; and (ii) rabbit is phylogenetically closer to primates than rodents. A. thaliana is an excel- lent plant model for which large molecular genomics tools and genetic resources are available. Furthermore, in the three systems, transitions between totipotency, multipotency and terminal differentiation can be ex- ploited and used to explore nuclear reorganization in relation to modifications of the pattern of gene expres- sion.

Pre-implantation rabbit embryos, the first system, of- fer the possibility to follow directly a differentiation process within the same intact organism. After the re- programmation of the parental genomes at fertilization, the first totipotent zygote leads in 5 days to a blastocyst where two different populations of cells coexist, already differentiated trophectoderm cells and totipotent cells of the inner cell mass. The whole process can take place in vitro in an appropriate culture medium. Finely tuned modifications of gene expression occur during this pe- riod, together with large restructuring of the nuclear ar- chitecture. The advantage of the rabbit is that zygotic genome activation occurs over an extended period of time as compared to the mouse [88].

The second system, the rabbit mammary gland, is subjected to successive cycles of intense development and involution which extend from gestation through lac- tation to weaning, and thus represents a powerful sys- tem to explore the impacts of physiological situations on genome function. During each cycle, genes coding

for milk proteins are turned on and off, together with many other genes. However, no studies of the nuclear organization of these epithelial cells in their tissue envi- ronment exist so far, only few data deal with mammary cell lines [77,89,90].

A. thaliana plantlets offer a third interesting system, since after digestion of the cell walls, totipotent “pro- toplasts” can be easily obtained. Upon culture in an appropriate medium, protoplasts can redifferentiate into tissues and new plants. A large reorganization of hete- rochromatin accompanies the first steps of dedifferenti- ation/redifferentiation in this system [73].

3.3. Development of modeling and computational tools To explore the nucleus on a global scale, new tools are required to establish computational models and to unravel principles governing the spatial distribution of nuclear compartments. Indeed most analyses were done so far in 2D, and very rarely in 3D [31,39,91,92].

Few quantitative measures have been performed [93, 94]. Standard quantitative analyses of the nuclear orga- nization are essentially based on distances measured on 2D images or on projections of 3D images, and rarely obtained directly in 3D. Distances can be measured between points (centers of compartments), or between points and surfaces (e.g. the nuclear envelope). Some- times distances are digitized into classes such as, for example, in the so-called erosion analysis used to an- alyze radial distributions within the nucleus [31].

The testing of complete spatial randomness is com- monly used to show that the nucleus is indeed spatially organized. A typical approach consists in choosing arbi- trary parameters (distance or angle) and comparing their experimental values with those obtained on patterns simulated according complete spatial randomness. Such an approach based on angles was used to test whether chromosome territories are located independently from each other [27]. In domains such as forestry, ecology, or epidemiology, generic statistical tools have been de- veloped for the analysis of spatial point patterns [95–

97]. To date, these tools have only rarely been used to investigate nuclear spatial organizations [98–101]. For example, Beil et al. [101] used the K-function to test whether chromocenters are completely random and to compare chromocenters patterns in undifferentiated and differentiated NB4 human cells. However, the conven- tional forms of spatial point pattern tools encompass no normalization for nuclear shape variations and are not applicable to repeated experiments.

To take into account the expected spatial hetero-

geneity of the nuclei and compare the heterochromatin

(8)

V. Gaudin et al. / C. R. Biologies 332 (2009) 937–946 943

(A)

(B)

Fig. 2. Spatial point patterns and analysis by distance functions.

A. The G-function is the cumulative distribution function of point to nearest point distances. Points are shown as black dots and are joined to their nearest neighbor by line segments. B. The F-function is the cumulative distribution function of arbitrary positions to nearest point distances. Sampled positions (shown as crosses) are connected to nearest points (black dots) by line segments.

distribution, we developed new tools based on process- ing and analysis of digital confocal images and a new statistical approach using variants of the spatial point patterns F- and G-functions (Fig. 2). These cumulative distribution functions of nearest-neighbor distances (G- function) or of distances between arbitrary points within the domain of interest (here the nucleus) and their near- est “events” (here nuclear elements) (F-function) in- deed provide more comprehensive descriptors than ra- dial distributions. Thus, they allow more powerful test- ing of hypotheses on spatial distributions. In this study, our F-function variant was the most appropriate tool and allowed us to highlight a non-random distribution of centric–pericentric heterochromatin in differentiated cells from our three biological systems.

The use of this F-function variant requires an adapted segmentation method to identify and localize the “in- teresting” biological objects within images collected on very different types of biological material. Furthermore, the segmentation procedure needs to be automated in or- der to analyze a large number of images. However, few algorithms are actually available as they may vary ac- cording to the specimen. Simple software, especially the commercially available ones, used to extract and calcu-

late 3D nuclear distributions can be used on cells grown in culture, with round shaped nuclei [27] but cannot be used for more sophisticated samples like irregularly shaped nuclei, tissue sections with clumped cells or low- contrasted fluorescent staining. For this project, we set up new automated or semi-automated segmentation al- gorithms adapted to the different nuclear compartments in the three dimensions of well-preserved nuclei.

4. Conclusion

The frame of the agroBI-INRA program entitled

“Nuclear Architecture: spatial modeling and application to a better understanding of differentiation/dedifferenti- ation mechanisms” allowed a fruitful confrontation be- tween animal and plant biologists, together with statis- ticians and modelers to define a common multidisci- plinary approach. The first results of this joint study will be presented in a forthcoming paper. We believe that comparisons between plant and animals will be highly beneficial to understand the similarities/differences in genome organization, gene regulation and cell differen- tiation processes.

Acknowledgements

This work was supported by an INRA agroBI Re- search Program (2006–2008) (http://www.inra.fr/

internet/Projets/agroBI/pageagroBI.html). We are grate- ful to Olivier Grandjean and Lionel Gissot, to the Lab- oratoire Commun de Cytologie (INRA Versailles), as well as to Pierre Adenot and the RIO MIMA2 platform (INRA Jouy-en-Josas) for technical support in confo- cal imaging. We thank Eric Biot, Nicole Houba-Hérin, Cathy Hue-Beauvais, Gaetan Lehman, Leila Tirichine, Liu Zichuan for biological experiments and develop- ment of image treatment and analysis tools. We thank Professor M.A. Ferguson-Smith and P. O’Brien (Cam- bridge Resource Centre for Comparative Genomics) for the gift of the rabbit chromosome-specific paints. We are indebted to Christophe Escudé for the LNA telom- eric probe and Federico Tessadori for providing images (Figs. 1I, 1K). We thank the Région Ile-de-France for the funding of the Leica TCS SP2 confocal microscope (INRA Versailles) and the Zeiss AxioObserver imaging Apotome system (INRA, Jouy).

References

[1] S. Gorski, T. Misteli, Systems biology in the cell nucleus, J. Cell Sci. 118 (2005) 4083–4092.

[2] T. Misteli, Beyond the sequence: Cellular organization of genome function, Cell 128 (2007) 787–800.

(9)

[3] T. Misteli, Concepts in nuclear architecture, Bioessays 27 (2005) 477–487.

[4] S.I. Grewal, S. Jia, Heterochromatin revisited, Nat. Rev.

Genet. 8 (2007) 35–46.

[5] F. Hediger, S.M. Gasser, Heterochromatin protein 1: Don’t judge the book by its cover! Curr. Opin. Genet. Dev. 16 (2006) 143–150.

[6] P. Dimitri, R. Caizzi, E. Giordano, M. Carmela Accardo, G. Lat- tanzi, G. Biamonti, Constitutive heterochromatin: A surprising variety of expressed sequences, Chromosoma 118 (2009) 419–

435.

[7] H. Bouzinba-Segard, A. Guais, C. Francastel, Accumulation of small murine minor satellite transcripts leads to impaired centromeric architecture and function, Proc. Natl. Acad. Sci.

USA 103 (2006) 8709–8714.

[8] I. Djupedal, K. Ekwall, Epigenetics: Heterochromatin meets RNAi, Cell Res. 19 (2009) 282–295.

[9] M. Carchilan, M. Delgado, T. Ribeiro, P. Costa-Nunes, A. Ca- perta, L. Morais-Cecilio, R.N. Jones, W. Viegas, A. Houben, Transcriptionally active heterochromatin in rye B chromo- somes, Plant Cell 19 (2007) 1738–1749.

[10] N. Gilbert, S. Boyle, H. Fiegler, K. Woodfine, N.P. Carter, W.A.

Bickmore, Chromatin architecture of the human genome: Gene- rich domains are enriched in open chromatin fibers, Cell 118 (2004) 555–566.

[11] I. Alcobia, R. Dilao, L. Parreira, Spatial associations of cen- tromeres in the nuclei of hematopoietic cells: Evidence for cell- type-specific organizational patterns, Blood 95 (2000) 1608–

1615.

[12] I. Alcobia, A.S. Quina, H. Neves, N. Clode, L. Parreira, The spatial organization of centromeric heterochromatin during nor- mal human lymphopoiesis: Evidence for ontogenically deter- mined spatial patterns, Exp. Cell Res. 290 (2003) 358–369.

[13] P. Fransz, J.H. De Jong, M. Lysak, M.R. Castiglione, I. Schu- bert, Interphase chromosomes in Arabidopsis are organized as well defined chromocenters from which euchromatin loops em- anate, Proc. Natl. Acad. Sci. USA 99 (2002) 14584–14589.

[14] S. de Nooijer, J. Wellink, B. Mulder, T. Bisseling, Non-specific interactions are sufficient to explain the position of heterochro- matic chromocenters and nucleoli in interphase nuclei, Nucleic Acids Res. (2009) 3558–3568.

[15] P.B. Talbert, R. Masuelli, A.P. Tyagi, L. Comai, S. Henikoff, Centromeric localization and adaptive evolution of an Ara- bidopsis histone H3 variant, Plant Cell 14 (2002) 1053–1066.

[16] Y. Fang, D.L. Spector, Centromere positioning and dynamics in living Arabidopsis plants, Mol. Biol. Cell 16 (2005) 5710–

5718.

[17] T. Cremer, C. Cremer, Chromosome territories, nuclear ar- chitecture and gene regulation in mammalian cells, Nat. Rev.

Genet. 2 (2001) 292–301.

[18] T. Cremer, M. Cremer, S. Dietzel, S. Muller, I. Solovei, S.

Fakan, Chromosome territories – a functional nuclear land- scape, Curr. Opin. Cell Biol. 18 (2006) 307–316.

[19] A. Berr, I. Schubert, Interphase chromosome arrangement in Arabidopsis thaliana is similar in differentiated and meristem- atic tissues and shows a transient mirror symmetry after nuclear division, Genetics 176 (2007) 853–863.

[20] A. Pecinka, V. Schubert, A. Meister, G. Kreth, M. Klatte, M.A.

Lysak, J. Fuchs, I. Schubert, Chromosome territory arrange- ment and homologous pairing in nuclei of Arabidopsis thaliana are predominantly random except for NOR-bearing chromo- somes, Chromosoma 113 (2004) 258–269.

[21] J. Ferreira, G. Paolella, C. Ramos, A.I. Lamond, Spatial organi- zation of large-scale chromatin domains in the nucleus: A mag- nified view of single chromosome territories, J. Cell Biol. 139 (1997) 1597–1610.

[22] J.A. Croft, J.M. Bridger, S. Boyle, P. Perry, P. Teague, W.A.

Bickmore, Differences in the localization and morphology of chromosomes in the human nucleus, J. Cell Biol. 145 (1999) 1119–1131.

[23] H. Tanabe, F.A. Habermann, I. Solovei, M. Cremer, T. Cremer, Non-random radial arrangements of interphase chromosome territories: Evolutionary considerations and functional implica- tions, Mutat. Res. 504 (2002) 37–45.

[24] H.A. Foster, L.R. Abeydeera, D.K. Griffin, J.M. Bridger, Non- random chromosome positioning in mammalian sperm nuclei, with migration of the sex chromosomes during late spermato- genesis, J. Cell Sci. 118 (2005) 1811–1820.

[25] N. Sadoni, S. Langer, C. Fauth, G. Bernardi, T. Cremer, B.M.

Turner, D. Zink, Nuclear organization of mammalian genomes.

Polar chromosome territories build up functionally distinct higher order compartments, J. Cell Biol. 146 (1999) 1211–

1226.

[26] R. Mayer, A. Brero, J. von Hase, T. Schroeder, T. Cremer, S.

Dietzel, Common themes and cell type specific variations of higher order chromatin arrangements in the mouse, BMC Cell Biol. 6 (2005) 44.

[27] A. Bolzer, G. Kreth, I. Solovei, D. Koehler, K. Saracoglu, C.

Fauth, S. Muller, R. Eils, C. Cremer, M.R. Speicher, T. Cremer, Three-dimensional maps of all chromosomes in human male fi- broblast nuclei and prometaphase rosettes, PLoS Biol. 3 (2005) e157.

[28] F.A. Habermann, M. Cremer, J. Walter, G. Kreth, J. von Hase, K. Bauer, J. Wienberg, C. Cremer, T. Cremer, I. Solovei, Ar- rangements of macro- and microchromosomes in chicken cells, Chromosome Res. 9 (2001) 569–584.

[29] O. Alexandrova, I. Solovei, T. Cremer, C.N. David, Replica- tion labeling patterns and chromosome territories typical of mammalian nuclei are conserved in the early metazoan Hydra, Chromosoma 112 (2003) 190–200.

[30] K. Kupper, A. Kolbl, D. Biener, S. Dittrich, J. von Hase, T.

Thormeyer, H. Fiegler, N.P. Carter, M.R. Speicher, T. Cremer, M. Cremer, Radial chromatin positioning is shaped by local gene density, not by gene expression, Chromosoma 116 (2007) 285–306.

[31] D. Zink, M.D. Amaral, A. Englmann, S. Lang, L.A. Clarke, C. Rudolph, F. Alt, K. Luther, C. Braz, N. Sadoni, J. Rose- necker, D. Schindelhauer, Transcription-dependent spatial ar- rangements of CFTR and adjacent genes in human cell nuclei, J. Cell Biol. 166 (2004) 815–825.

[32] S.T. Kosak, J.A. Skok, K.L. Medina, R. Riblet, M.M. Le Beau, A.G. Fisher, H. Singh, Subnuclear compartmentalization of immunoglobulin loci during lymphocyte development, Sci- ence 296 (2002) 158–162.

[33] R.R. Williams, V. Azuara, P. Perry, S. Sauer, M. Dvorkina, H.

Jorgensen, J. Roix, P. McQueen, T. Misteli, M. Merkenschlager, A.G. Fisher, Neural induction promotes large-scale chromatin reorganisation of the Mash1 locus, J. Cell Sci. 119 (2006) 132–

140.

[34] S. Dietzel, K. Zolghadr, C. Hepperger, A.S. Belmont, Differ- ential large-scale chromatin compaction and intranuclear posi- tioning of transcribed versus non-transcribed transgene arrays containing beta-globin regulatory sequences, J. Cell Sci. 117 (2004) 4603–4614.

(10)

V. Gaudin et al. / C. R. Biologies 332 (2009) 937–946 945

[35] J.H. Brickner, P. Walter, Gene recruitment of the activated INO1 locus to the nuclear membrane, PLoS Biol. 2 (2004) e342.

[36] J.M. Casolari, C.R. Brown, S. Komili, J. West, H. Hieronymus, P.A. Silver, Genome-wide localization of the nuclear transport machinery couples transcriptional status and nuclear organiza- tion, Cell 117 (2004) 427–439.

[37] Y. Gruenbaum, A. Margalit, R.D. Goldman, D.K. Shumaker, K.L. Wilson, The nuclear lamina comes of age, Nat. Rev. Mol.

Cell Biol. 6 (2005) 21–31.

[38] M.O. Scheuermann, J. Tajbakhsh, A. Kurz, K. Saracoglu, R.

Eils, P. Lichter, Topology of genes and nontranscribed se- quences in human interphase nuclei, Exp. Cell Res. 301 (2004) 266–279.

[39] A. Kurz, S. Lampel, J.E. Nickolenko, J. Bradl, A. Benner, R.M.

Zirbel, T. Cremer, P. Lichter, Active and inactive genes localize preferentially in the periphery of chromosome territories, J. Cell Biol. 135 (1996) 1195–1205.

[40] I. Faro-Trindade, P.R. Cook, Transcription factories: Structures conserved during differentiation and evolution, Biochem. Soc.

Trans. 34 (2006) 1133–1137.

[41] H. Albiez, M. Cremer, C. Tiberi, L. Vecchio, L. Schermelleh, S. Dittrich, K. Kupper, B. Joffe, T. Thormeyer, J. von Hase, S.

Yang, K. Rohr, H. Leonhardt, I. Solovei, C. Cremer, S. Fakan, T. Cremer, Chromatin domains and the interchromatin com- partment form structurally defined and functionally interacting nuclear networks, Chromosome Res. 14 (2006) 707–733.

[42] R.R. Williams, S. Broad, D. Sheer, J. Ragoussis, Subchromoso- mal positioning of the epidermal differentiation complex (EDC) in keratinocyte and lymphoblast interphase nuclei, Exp. Cell Res. 272 (2002) 163–175.

[43] E.V. Volpi, E. Chevret, T. Jones, R. Vatcheva, J. Williamson, S.

Beck, R.D. Campbell, M. Goldsworthy, S.H. Powis, J. Ragous- sis, J. Trowsdale, D. Sheer, Large-scale chromatin organization of the major histocompatibility complex and other regions of human chromosome 6 and its response to interferon in inter- phase nuclei, J. Cell Sci. 113 (2000) 1565–1576 (Pt 9).

[44] N.L. Mahy, P.E. Perry, S. Gilchrist, R.A. Baldock, W.A. Bick- more, Spatial organization of active and inactive genes and non- coding DNA within chromosome territories, J. Cell Biol. 157 (2002) 579–589.

[45] P.J. Verschure, I. Van Der Kraan, J.M. Enserink, M.J. Mone, E.M. Manders, R. Van Driel, Large-scale chromatin organiza- tion and the localization of proteins involved in gene expression in human cells, J. Histochem. Cytochem. 50 (2002) 1303–1312.

[46] E. Heard, W. Bickmore, The ins and outs of gene regulation and chromosome territory organisation, Curr. Opin. Cell Biol. 19 (2007) 311–316.

[47] K.E. Brown, S.S. Guest, S.T. Smale, K. Hahm, M. Merken- schlager, A.G. Fisher, Association of transcriptionally silent genes with Ikaros complexes at centromeric heterochromatin, Cell 91 (1997) 845–854.

[48] C. Francastel, W. Magis, M. Groudine, Nuclear relocation of a transactivator subunit precedes target gene activation, Proc.

Natl. Acad. Sci. USA 98 (2001) 12120–12125.

[49] S.T. Kosak, M. Groudine, Gene order and dynamic domains, Science 306 (2004) 644–647.

[50] S. Chambeyron, W.A. Bickmore, Does looping and cluster- ing in the nucleus regulate gene expression? Curr. Opin. Cell Biol. 16 (2004) 256–262.

[51] S.H. Kim, P.G. McQueen, M.K. Lichtman, E.M. Shevach, L.A.

Parada, T. Misteli, Spatial genome organization during T-cell differentiation, Cytogenet. Genome Res. 105 (2004) 292–301.

[52] R.C. Su, K.E. Brown, S. Saaber, A.G. Fisher, M. Merken- schlager, S.T. Smale, Dynamic assembly of silent chromatin during thymocyte maturation, Nat. Genet. 36 (2004) 502–506.

[53] C.S. Osborne, L. Chakalova, J.A. Mitchell, A. Horton, A.L.

Wood, D.J. Bolland, A.E. Corcoran, P. Fraser, Myc dynamically and preferentially relocates to a transcription factory occupied by Igh, PLoS Biol. 5 (2007) e192.

[54] J.M. Brown, J. Leach, J.E. Reittie, A. Atzberger, J. Lee- Prudhoe, W.G. Wood, D.R. Higgs, F.J. Iborra, V.J. Buckle, Coregulated human globin genes are frequently in spatial prox- imity when active, J. Cell Biol. 172 (2006) 177–187.

[55] I. Raska, P.J. Shaw, D. Cmarko, Structure and function of the nucleolus in the spotlight, Curr. Opin. Cell Biol. 18 (2006) 325–

334.

[56] Q. Hu, Y.S. Kwon, E. Nunez, M.D. Cardamone, K.R. Hutt, K.A. Ohgi, I. Garcia-Bassets, D.W. Rose, C.K. Glass, M.G.

Rosenfeld, X.D. Fu, Enhancing nuclear receptor-induced tran- scription requires nuclear motor and LSD1-dependent gene networking in interchromatin granules, Proc. Natl. Acad. Sci.

USA 105 (2008) 19199–19204.

[57] S.A. Lelievre, V.M. Weaver, J.A. Nickerson, C.A. Larabell, A.

Bhaumik, O.W. Petersen, M.J. Bissell, Tissue phenotype de- pends on reciprocal interactions between the extracellular ma- trix and the structural organization of the nucleus, Proc. Natl.

Acad. Sci. USA 95 (1998) 14711–14716.

[58] D. Koehler, V. Zakhartchenko, L. Froenicke, G. Stone, R.

Stanyon, E. Wolf, T. Cremer, A. Brero, Changes of higher or- der chromatin arrangements during major genome activation in bovine preimplantation embryos, Exp. Cell Res. (2009) 2053–

2063.

[59] S. Stadler, V. Schnapp, R. Mayer, S. Stein, C. Cremer, C.

Bonifer, T. Cremer, S. Dietzel, The architecture of chicken chromosome territories changes during differentiation, BMC Cell Biol. 5 (2004) 44.

[60] E. Meshorer, T. Misteli, Chromatin in pluripotent embryonic stem cells and differentiation, Nat. Rev. Mol. Cell Biol. 7 (2006) 540–546.

[61] A. Brero, H.P. Easwaran, D. Nowak, I. Grunewald, T. Cremer, H. Leonhardt, M.C. Cardoso, Methyl CpG-binding proteins in- duce large-scale chromatin reorganization during terminal dif- ferentiation, J. Cell Biol. 169 (2005) 733–743.

[62] M. Beil, D. Durschmied, S. Paschke, B. Schreiner, U. Nolte, A. Bruel, T. Irinopoulou, Spatial distribution patterns of inter- phase centromeres during retinoic acid-induced differentiation of promyelocytic leukemia cells, Cytometry 47 (2002) 217–

225.

[63] G. Martou, U. De Boni, Nuclear topology of murine, cerebellar Purkinje neurons: Changes as a function of development, Exp.

Cell Res. 256 (2000) 131–139.

[64] N. Chaly, S.B. Munro, Centromeres reposition to the nuclear periphery during L6E9 myogenesis in vitro, Exp. Cell Res. 223 (1996) 274–278.

[65] A.V. Probst, F. Santos, W. Reik, G. Almouzni, W. Dean, Structural differences in centromeric heterochromatin are spa- tially reconciled on fertilisation in the mouse zygote, Chromo- soma 116 (2007) 403–415.

[66] C. Martin, N. Beaujean, V. Brochard, C. Audouard, D. Zink, P. Debey, Genome restructuring in mouse embryos during re- programming and early development, Dev. Biol. 292 (2006) 317–332.

[67] V. Merico, J. Barbieri, M. Zuccotti, B. Joffe, T. Cremer, C.A.

Redi, I. Solovei, S. Garagna, Epigenomic differentiation in

(11)

mouse preimplantation nuclei of biparental, parthenote and cloned embryos, Chromosome Res. 15 (2007) 341–360.

[68] O. Mathieu, Z. Jasencakova, I. Vaillant, A.V. Gendrel, V. Colot, I. Schubert, S. Tourmente, Changes in 5S rDNA chromatin or- ganization and transcription during heterochromatin establish- ment in Arabidopsis, Plant Cell 15 (2003) 2929–2939.

[69] F. Tessadori, R. van Driel, P. Fransz, Cytogenetics as a tool to study gene regulation, Trends Plant Sci. 9 (2004) 147–153.

[70] F. Tessadori, R.K. Schulkes, R. van Driel, P. Fransz, Light- regulated large-scale reorganization of chromatin during the floral transition in Arabidopsis, Plant J. 50 (2007) 848–857.

[71] C. Baroux, A. Pecinka, J. Fuchs, I. Schubert, U. Grossniklaus, The triploid endosperm genome of Arabidopsis adopts a pecu- liar, parental-dosage-dependent chromatin organization, Plant Cell 19 (2007) 1782–1794.

[72] S. Costa, P. Shaw, Chromatin organization and cell fate switch respond to positional information in Arabidopsis, Nature 439 (2006) 493–496.

[73] F. Tessadori, M.C. Chupeau, Y. Chupeau, M. Knip, S. Germann, R. van Driel, P. Fransz, V. Gaudin, Large-scale dissociation and sequential reassembly of pericentric heterochromatin in ded- ifferentiated Arabidopsis cells, J. Cell Sci. 120 (2007) 1200–

1208.

[74] J. Mateos-Langerak, S. Goetze, H. Leonhardt, T. Cremer, R.

van Driel, C. Lanctot, Nuclear architecture: Is it important for genome function and can we prove it? J. Cell Biochem. 102 (2007) 1067–1075.

[75] G. Chandramouly, P.C. Abad, D.W. Knowles, S.A. Lelievre, The control of tissue architecture over nuclear organization is crucial for epithelial cell fate, J. Cell Sci. 120 (2007) 1596–

1606.

[76] K.J. Meaburn, T. Misteli, Locus-specific and activity- independent gene repositioning during early tumorigenesis, J. Cell Biol. 180 (2008) 39–50.

[77] P. Kaminker, C. Plachot, S.H. Kim, P. Chung, D. Crippen, O.W. Petersen, M.J. Bissell, J. Campisi, S.A. Lelievre, Higher- order nuclear organization in growth arrest of human mammary epithelial cells: A novel role for telomere-associated protein TIN2, J. Cell Sci. 118 (2005) 1321–1330.

[78] E. Chytilova, J. Macas, D.W. Galbraith, Green fluorescent pro- tein targeted to the nucleus, a transgenic phenotype useful for studies in plant biology, Annals of Botany 83 (1999) 645–654.

[79] N. Kato, E. Lam, Chromatin of endoreduplicated pavement cells has greater range of movement than that of diploid guard cells in Arabidopsis thaliana, J. Cell Sci. 116 (2003) 2195–

2201.

[80] D. Marenduzzo, I. Faro-Trindade, P.R. Cook, What are the molecular ties that maintain genomic loops? Trends Genet. 23 (2007) 126–133.

[81] J.M. O’Sullivan, D.M. Sontam, R. Grierson, B. Jones, Re- peated elements coordinate the spatial organization of the yeast genome, Yeast 26 (2009) 125–138.

[82] M. Dundr, U. Hoffmann-Rohrer, Q. Hu, I. Grummt, L.I. Roth- blum, R.D. Phair, T. Misteli, A kinetic framework for a mam- malian RNA polymerase in vivo, Science 298 (2002) 1623–

1626.

[83] R. Hancock, A role for macromolecular crowding effects in the assembly and function of compartments in the nucleus, J. Struct. Biol. 146 (2004) 281–290.

[84] D. Marenduzzo, K. Finan, P.R. Cook, The depletion attraction:

An underappreciated force driving cellular organization, J. Cell Biol. 175 (2006) 681–686.

[85] S. Asakura, F. Oosawa, Interactions between particles sus- pended in solutions of macromolecules, J. Polym. Sci. B 33 (1958) 183–192.

[86] M. Guenatri, D. Bailly, C. Maison, G. Almouzni, Mouse centric and pericentric satellite repeats form distinct functional hete- rochromatin, J. Cell Biol. 166 (2004) 493–505.

[87] A. Goncalves Dos Santos Silva, R. Sarkar, J. Harizanova, A.

Guffei, M. Mowat, Y. Garini, S. Mai, Centromeres in cell division, evolution, nuclear organization and disease, J. Cell Biochem. 104 (2008) 2040–2058.

[88] A. Brunet-Simon, G. Henrion, J.P. Renard, V. Duranthon, Onset of zygotic transcription and maternal transcript legacy in the rabbit embryo, Mol. Reprod. Dev. 58 (2001) 127–136.

[89] M.J. Bissell, V.M. Weaver, S.A. Lelievre, F. Wang, O.W. Pe- tersen, K.L. Schmeichel, Tissue structure, nuclear organization, and gene expression in normal and malignant breast, Cancer Res. 59 (1999) 1757–1763s; discussion 1763s–1764s.

[90] J.L. Salisbury, The contribution of epigenetic changes to ab- normal centrosomes and genomic instability in breast cancer, J. Mammary Gland Biol. Neoplasia 6 (2001) 203–212.

[91] M. Gue, J.S. Sun, T. Boudier, Simultaneous localization of MLL, AF4 and ENL genes in interphase nuclei by 3D-FISH:

MLL translocation revisited, BMC Cancer 6 (2006) 20.

[92] N.L. Mahy, P.E. Perry, W.A. Bickmore, Gene density and tran- scription influence the localization of chromatin outside of chromosome territories detectable by FISH, J. Cell Biol. 159 (2002) 753–763.

[93] O. Ronneberger, D. Baddeley, F. Scheipl, P.J. Verveer, H.

Burkhardt, C. Cremer, L. Fahrmeir, T. Cremer, B. Joffe, Spatial quantitative analysis of fluorescently labeled nuclear structures:

Problems, methods, pitfalls, Chromosome Res. 16 (2008) 523–

562.

[94] C. Shiels, N.M. Adams, S.A. Islam, D.A. Stephens, P.S.

Freemont, Quantitative analysis of cell nucleus organisation, PLoS Comput. Biol. 3 (2007) e138.

[95] A. Baddeley, P. Gregori, J. Mateu, R. Stoica, D. Stoyan, Case Studies in Spatial Point Process Modeling, Lecture Notes in Statistics, vol. 185, Springer-Verlag, 2006.

[96] P.J. Diggle, Statistical Analysis of Spatial Point Patterns, 2nd edition, A Hodder Arnold Publication, Oxford Univ. Press, London, 2003.

[97] N.A.C. Cressie, Statistics for Spatial Data, Series in Probability and Statistics, Wiley-Interscience, 1993.

[98] K.J. McManus, D.A. Stephens, N.M. Adams, S.A. Islam, P.S.

Freemont, M.J. Hendzel, The transcriptional regulator CBP has defined spatial associations within interphase nuclei, PLoS Comput. Biol. 2 (2006) e139.

[99] D.W. Young, S.K. Zaidi, P.S. Furcinitti, A. Javed, A.J. van Wij- nen, J.L. Stein, J.B. Lian, G.S. Stein, Quantitative signature for architectural organization of regulatory factors using intranu- clear informatics, J. Cell Sci. 117 (2004) 4889–4896.

[100] H.J. Noordmans, K. van der Kraan, R. van Driel, A.W. Smeul- ders, Randomness of spatial distributions of two proteins in the cell nucleus involved in mRNA synthesis and their relationship, Cytometry 33 (1998) 297–309.

[101] M. Beil, H. Braxmeier, F. Fleischer, V. Schmidt, P. Walther, Quantitative analysis of keratin filament networks in scanning electron microscopy images of cancer cells, J. Microsc. 220 (2005) 84–95.

Références

Documents relatifs

Virtual plant models are powerful tools to better understand growth and functioning of plants and their  interaction  with  their  environment  (light,  pest 

The OpenAlea architecture consist s of: (a) a set of tools to integrate heterogeneous models implemented in v ari ous languages and on different platforms ; (b) a component

Sequential processing using a triangular filter with Mel-scaling and a Hamming window is applied to increase the accuracy of the FB method, and the instability criterion is

Systematic combination of Euler poles available in the literature, in addition to our new set of data, shows that a wide range of Arabia-Sinai pole positions and angular

Compilation of new and published apatite fission track data along a transect of the west-central Pyrenees shows that exhumation across the Partial Annealing Zone (~120–60°C)

As mentioned above, the first application of our methodology is focused on the nuclear architec- ture in A. The nucleus is an ordered and complex organelle that contains several

Taking both expectations in mind, in this dissertation we have designed and investigated different kinds of nonlinear silicon plasmonic waveguides and exploited the

Then the mean squared error of those models is computed on the validation set (again the same for all models) and the nuisance parameters with the lowest error